Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 13(9): 820, 2022 09 24.
Article in English | MEDLINE | ID: mdl-36153321

ABSTRACT

Molecular understanding of osteogenic differentiation (OD) of human bone marrow-derived mesenchymal stem cells (hBMSCs) is important for regenerative medicine and has direct implications for cancer. We report that the RNF4 ubiquitin ligase is essential for OD of hBMSCs, and that RNF4-deficient hBMSCs remain as stalled progenitors. Remarkably, incubation of RNF4-deficient hBMSCs in conditioned media of differentiating hBMSCs restored OD. Transcriptional analysis of RNF4-dependent gene signatures identified two secreted factors that act downstream of RNF4 promoting OD: (1) BMP6 and (2) the BMP6 co-receptor, RGMb (Dragon). Indeed, knockdown of either RGMb or BMP6 in hBMSCs halted OD, while only the combined co-addition of purified RGMb and BMP6 proteins to RNF4-deficient hBMSCs fully restored OD. Moreover, we found that the RNF4-RGMb-BMP6 axis is essential for survival and tumorigenicity of osteosarcoma and therapy-resistant melanoma cells. Importantly, patient-derived sarcomas such as osteosarcoma, Ewing sarcoma, liposarcomas, and leiomyosarcomas exhibit high levels of RNF4 and BMP6, which are associated with reduced patient survival. Overall, we discovered that the RNF4~BMP6~RGMb axis is required for both OD and tumorigenesis.


Subject(s)
Bone Morphogenetic Protein 6 , Cell Adhesion Molecules, Neuronal , Osteogenesis , Osteosarcoma , Transcription Factors , Bone Marrow Cells/metabolism , Bone Morphogenetic Protein 6/metabolism , Cell Adhesion Molecules, Neuronal/metabolism , Cell Differentiation , Cell Survival , Cells, Cultured , Culture Media, Conditioned/metabolism , Humans , Ligases/metabolism , Nuclear Proteins/metabolism , Osteosarcoma/metabolism , Transcription Factors/metabolism , Ubiquitins/metabolism
2.
Cells ; 10(9)2021 09 09.
Article in English | MEDLINE | ID: mdl-34572023

ABSTRACT

A hallmark of cancer is dysregulated protein turnover (proteostasis), which involves pathologic ubiquitin-dependent degradation of tumor suppressor proteins, as well as increased oncoprotein stabilization. The latter is due, in part, to mutation within sequences, termed degrons, which are required for oncoprotein recognition by the substrate-recognition enzyme, E3 ubiquitin ligase. Stabilization may also result from the inactivation of the enzymatic machinery that mediates the degradation of oncoproteins. Importantly, inactivation in cancer of E3 enzymes that regulates the physiological degradation of oncoproteins, results in tumor cells that accumulate multiple active oncoproteins with prolonged half-lives, leading to the development of "degradation-resistant" cancer cells. In addition, specific sequences may enable ubiquitinated proteins to evade degradation at the 26S proteasome. While the ubiquitin-proteasome pathway was originally discovered as central for protein degradation, in cancer cells a ubiquitin-dependent protein stabilization pathway actively translates transient mitogenic signals into long-lasting protein stabilization and enhances the activity of key oncoproteins. A central enzyme in this pathway is the ubiquitin ligase RNF4. An intimate link connects protein stabilization with tumorigenesis in experimental models as well as in the clinic, suggesting that pharmacological inhibition of protein stabilization has potential for personalized medicine in cancer. In this review, we highlight old observations and recent advances in our knowledge regarding protein stabilization.


Subject(s)
Ubiquitin/metabolism , Humans , Nuclear Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Signal Transduction/physiology , Ubiquitin-Protein Ligases/metabolism
3.
Elife ; 102021 02 25.
Article in English | MEDLINE | ID: mdl-33629655

ABSTRACT

A hallmark of aging is loss of differentiated cell identity. Aged Drosophila midgut differentiated enterocytes (ECs) lose their identity, impairing tissue homeostasis. To discover identity regulators, we performed an RNAi screen targeting ubiquitin-related genes in ECs. Seventeen genes were identified, including the deubiquitinase Non-stop (CG4166). Lineage tracing established that acute loss of Non-stop in young ECs phenocopies aged ECs at cellular and tissue levels. Proteomic analysis unveiled that Non-stop maintains identity as part of a Non-stop identity complex (NIC) containing E(y)2, Sgf11, Cp190, (Mod) mdg4, and Nup98. Non-stop ensured chromatin accessibility, maintaining the EC-gene signature, and protected NIC subunit stability. Upon aging, the levels of Non-stop and NIC subunits declined, distorting the unique organization of the EC nucleus. Maintaining youthful levels of Non-stop in wildtype aged ECs safeguards NIC subunits, nuclear organization, and suppressed aging phenotypes. Thus, Non-stop and NIC, supervise EC identity and protects from premature aging.


Subject(s)
Aging, Premature/genetics , Aging/genetics , Drosophila Proteins/genetics , Drosophila melanogaster/physiology , Enterocytes/physiology , Animals , Disease Models, Animal , Drosophila Proteins/metabolism , Female , Male , Phenotype , Proteome
4.
Cell Mol Life Sci ; 78(7): 3141-3158, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33507327

ABSTRACT

Regulation of the differentiated identity requires active and continued supervision. Inability to maintain the differentiated state is a hallmark of aging and aging-related disease. To maintain cellular identity, a network of nuclear regulators is devoted to silencing previous and non-relevant gene programs. This network involves transcription factors, epigenetic regulators, and the localization of silent genes to heterochromatin. Together, identity supervisors mold and maintain the unique nuclear environment of the differentiated cell. This review describes recent discoveries regarding mechanisms and regulators that supervise the differentiated identity and protect from de-differentiation, tumorigenesis, and attenuate forced somatic cell reprograming. The review focuses on mechanisms involved in H3K9me3-decorated heterochromatin and the importance of nuclear lamins in cell identity. We outline how the biophysical properties of these factors are involved in self-compartmentalization of heterochromatin and cell identity. Finally, we discuss the relevance of these regulators to aging and age-related disease.


Subject(s)
Aging/pathology , Cell Differentiation , Cell Nucleus/physiology , Cellular Reprogramming , Neoplasms/pathology , Animals , Heterochromatin , Histones , Humans , Nuclear Lamina
5.
Elife ; 82019 07 16.
Article in English | MEDLINE | ID: mdl-31310235

ABSTRACT

The inability of differentiated cells to maintain their identity is a hallmark of age-related diseases. We found that the transcription factor Hey supervises the identity of differentiated enterocytes (ECs) in the adult Drosophila midgut. Lineage tracing established that Hey-deficient ECs are unable to maintain their unique nuclear organization and identity. To supervise cell identity, Hey determines the expression of nuclear lamins, switching from a stem-cell lamin configuration to a differentiated lamin configuration. Moreover, continued Hey expression is required to conserve large-scale nuclear organization. During aging, Hey levels decline, and EC identity and gut homeostasis are impaired, including pathological reprograming and compromised gut integrity. These phenotypes are highly similar to those observed upon acute targeting of Hey or perturbation of lamin expression in ECs in young adults. Indeed, aging phenotypes were suppressed by continued expression of Hey in ECs, suggesting that a Hey-lamin network safeguards nuclear organization and differentiated cell identity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Drosophila Proteins/metabolism , Drosophila/physiology , Enterocytes/physiology , Lamins/metabolism , Aging/pathology , Animals , Stem Cells/physiology
6.
Bio Protoc ; 9(22): e3435, 2019 Nov 20.
Article in English | MEDLINE | ID: mdl-33654931

ABSTRACT

Regulation of gene expression involves dynamic changes in chromatin organization, where in many cases open chromatin structure correlates with gene activation. Several methods enable monitoring changes in chromatin accessibility, including ATAC-seq, FAIRE-seq, MNase-seq and DNAse-seq methods, which involve Next-generation-sequencing (NGS). Focusing on the adult Drosophila differentiated gut enterocytes (ECs) we used a sequencing-free method that enables visualizing and semi-quantifying large-scale changes in chromatin structure using in vitro methylation assay with the bacterial CpG Methyltransferase, M. Sssl, that determine chromatin accessibility. In brief, as CpG methylation is minimal in differentiated somatic Drosophila cells, we used the bacterial M. SssI enzyme to methylate CpG dinucleotides in situ depending on their chromatin accessibility. The methylated dinucleotides are detected using 5mCytosine monoclonal antibody and nuclei are visualized microscopically. Thus, the 5mC method enables to monitor large-scale chromatin changes in heterogenic cellular tissue focusing on the cell type of interest and without the need for cell purification or NGS.

7.
Cells ; 7(10)2018 Sep 26.
Article in English | MEDLINE | ID: mdl-30261639

ABSTRACT

The HECT-type ubiquitin ligase HECT, UBA and WWE Domain Containing 1, (HUWE1) regulates key cancer-related pathways, including the Myc oncogene. It affects cell proliferation, stress and immune signaling, mitochondria homeostasis, and cell death. HUWE1 is evolutionarily conserved from Caenorhabditis elegance to Drosophila melanogaster and Humans. Here, we report that the Drosophila ortholog, dHUWE1 (CG8184), is an essential gene whose loss results in embryonic lethality and whose tissue-specific disruption establishes its regulatory role in larval salivary gland development. dHUWE1 is essential for endoreplication of salivary gland cells and its knockdown results in the inability of these cells to replicate DNA. Remarkably, dHUWE1 is a survival factor that prevents premature activation of JNK signaling, thus preventing the disintegration of the salivary gland, which occurs physiologically during pupal stages. This function of dHUWE1 is general, as its inhibitory effect is observed also during eye development and at the organismal level. Epistatic studies revealed that the loss of dHUWE1 is compensated by dMyc proeitn expression or the loss of dmP53. dHUWE1 is therefore a conserved survival factor that regulates organ formation during Drosophila development.

8.
J Dev Biol ; 6(1)2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29615551

ABSTRACT

The ubiquitin and SUMO (small ubiquitin-like modifier) pathways modify proteins that in turn regulate diverse cellular processes, embryonic development, and adult tissue physiology. These pathways were originally discovered biochemically in vitro, leading to a long-standing challenge of elucidating both the molecular cross-talk between these pathways and their biological importance. Recent discoveries in Drosophila established that ubiquitin and SUMO pathways are interconnected via evolutionally conserved SUMO-targeted ubiquitin ligase (STUbL) proteins. STUbL are RING ubiquitin ligases that recognize SUMOylated substrates and catalyze their ubiquitination, and include Degringolade (Dgrn) in Drosophila and RNF4 and RNF111 in humans. STUbL are essential for early development of both the fly and mouse embryos. In the fly embryo, Dgrn regulates early cell cycle progression, sex determination, zygotic gene transcription, segmentation, and neurogenesis, among other processes. In the fly adult, Dgrn is required for systemic immune response to pathogens and intestinal stem cell regeneration upon infection. These functions of Dgrn are highly conserved in humans, where RNF4-dependent ubiquitination potentiates key oncoproteins, thereby accelerating tumorigenesis. Here, we review the lessons learned to date in Drosophila and highlight their relevance to cancer biology.

9.
Fly (Austin) ; 5(4): 340-4, 2011.
Article in English | MEDLINE | ID: mdl-21857164

ABSTRACT

Posttranscriptional modifications of proteins by the ubiquitin and SUMO (Small Ubiquitin-related Modifier) pathways regulate the function of protein networks, enable cells to respond to signaling cues during development, and to cope with the changing environment during adult life. Both modifications can impact protein stability, localization, protein-protein interactions and/or function. While both pathways have been well studied individually, the long-speculated nature of crosstalk between SUMO and ubiquitin pathways has been molecularly enigmatic. Recent work in yeast and mammalian cells identified the connection between the two pathways in the form of a conserved family of RING finger ubiquitin ligases termed SUMO-Targeted ubiquitin ligases (STUbLs). These proteins bind to SUMOylated substrates via their SUMO interaction motif and subsequently target them for ubiquitylation. Characterization of Degringolade (Dgrn), a STUbL gene in the fly genome, enabled us to evaluate the contribution of STUbLs to the development of multi-cellular organisms. Analysis of dgrn mutants showed that they are required for cyto-nuclear organization during early embryonic development, and are likely required to cope with mitotic stress and DNA damage. Furthermore, in transcription, STUbLs regulate protein-protein interactions, and are part of molecular machinery that regulates co-repressor choice and gene-expression selectivity during development.


Subject(s)
Drosophila Proteins/physiology , Drosophila/enzymology , Models, Biological , Small Ubiquitin-Related Modifier Proteins/physiology , Ubiquitin-Protein Ligases/physiology , Animals , Drosophila/embryology , Drosophila/metabolism , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Embryo, Nonmammalian , Embryonic Development/genetics , Gene Expression Regulation, Developmental , Sumoylation , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
SELECTION OF CITATIONS
SEARCH DETAIL
...